Abstract

Indoleamine 2,3-dioxygenase 1 (IDO1) is a single chain oxidoreductase that catalyzes tryptophan degradation to kynurenine. In cancer, it exerts an immunosuppressive function as part of an acquired mechanism of immune escape. Recently, we demonstrated that IDO1 expression is significantly higher in all thyroid cancer histotypes compared with normal thyroid and that its expression levels correlate with T regulatory (Treg) lymphocyte densities in the tumor microenvironment. BRAFV600E- and RET/PTC3-expressing PcCL3 cells were used as cellular models for the evaluation of IDO1 expression in thyroid carcinoma cells and for the study of involved signal transduction pathways. BRAFV600E-expressing PcCL3 cells did not show IDO1 expression. Conversely, RET/PTC3-expressing cells were characterized by a high IDO1 expression. Moreover, we found that, the STAT1-IRF1 pathway was instrumental for IDO1 expression in RET/PTC3 expressing cells. In detail, RET/PTC3 induced STAT1 overexpression and phosphorylation at Ser-727 and Tyr-701. STAT1 transcriptional regulation appeared to require activation of the canonical NF-κB pathway. Conversely, activation of the MAPK and PI3K-AKT pathways primarily regulated Ser-727 phosphorylation, whereas a physical interaction between RET/PTC3 and STAT1, followed by a direct tyrosine phosphorylation event, was necessary for STAT1 Tyr-701 phosphorylation. These data provide the first evidence of a direct link between IDO1 expression and the oncogenic activation of RET in thyroid carcinoma and describe the involved signal transduction pathways. Moreover, they suggest possible novel molecular targets for the abrogation of tumor microenvironment immunosuppression. The detection of those targets is becoming increasingly important to yield the full function of novel immune checkpoint inhibitors.

Highlights

  • BRAF point mutations and RET gene rearrangements (RET/ PTCs) are the two major groups of mutations involved in papillary thyroid carcinoma initiation and progression

  • We demonstrated that IDO1 expression is significantly higher in all thyroid cancer histotypes compared with normal thyroid and that its expression levels correlate with T regulatory (Treg) lymphocyte densities in the thyroid tumor microenvironment [7]

  • RET/PTC3-induced IDO1 Expression in PTC3–5 Cells Depends on Activation of a RET/PTC3-STAT1-IRF1 Pathway— The dissection of RET/PTC3-activated signaling pathways involved in the induction of IDO1 expression focused on STAT1, a transcription factor known to play a central role in

Read more

Summary

Introduction

BRAF point mutations and RET gene rearrangements (RET/ PTCs) are the two major groups of mutations involved in papillary thyroid carcinoma initiation and progression. In PTC3–5 cells treated with Dox for up to 96 h, characterized by robust IDO1 mRNA overexpression, RET/PTC3 induced both up-regulation of STAT1 protein expression and its full activation through the phosphorylation of tyrosine 701 and serine 727 (Fig. 2). IDO1 expression on RET/PTC3-induced STAT1 activation, we performed an RNAi experiment.

Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call